Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

VIRAL IMMUNOLOGY

Volume 00, Number 00, 2020


ª Mary Ann Liebert, Inc.
Pp. 1–11
DOI: 10.1089/vim.2020.0082

An Improved DNA Vaccine Against Bovine Herpesvirus-1


Using CD40L and a Chemical Adjuvant Induces Specific
Cytotoxicity in Mice

Cecilia A. Langellotti,1,* Mariela Gammella,1,* Ivana Soria,1 Carolina Bellusci,2 Valeria Quattrocchi,1
Monica Vermeulen,3 Claudia Mongini,1 and Patricia I. Zamorano4
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

Abstract

Bovine herpesvirus-1 (BoHV-1) uses many mechanisms to elude the immune system; one of them is spreading
intracellularly, even in the presence of specific antiviral antibodies. Cytotoxic T lymphocytes (CTLs) are
necessary to eliminate the virus. The main preventive strategy is vaccination based on inactivated virus. These
vaccines are poor inducers of cellular immune responses, and complicate serological diagnosis and determi-
nation of the real prevalence of infection. DNA vaccines are a good option because of the capacity of
Differentiating Infected from Vaccinated Animals—(DIVA vaccine)—and may be the best way to induce
cytotoxic responses. Although this type of vaccines leads to only weak ‘‘in vivo’’ expression and poor immune
responses, incorporation of molecular and/or chemical adjuvants can improve the latter, both in magnitude
and in direction. In this study, we have investigated the specific immune responses elicited in mice by DNA
vaccines based on the BoHV-1 glycoprotein D (pCIgD) with and without two different adjuvants: a plasmid
encoding for murine CD40L (pCD40L) or Montanide 1113101PR (101). Mice vaccinated with
pCIgD+CD40L, pCIgD+101, and pCIgD+CD40L+101 developed significantly higher specific antibody titers
against BoHV-1 than the pCIgD group ( p < 0.01). The animals vaccinated with pCgD+pCD40L+101 raised
significantly higher levels of IgG2a and IgG2b ( p < 0.01 and p < 0.001, respectively) than mice vaccinated with
pCIgD alone. On the contrary, when the activity of CTL against cells infected with BoHV-1 was measured, the
vaccine pCgD+pCD40L+101 induced significantly higher levels of cytotoxicity activity ( p < 0.001) than pCIgD
alone. A significant increase in the CD4+ populations in the group receiving pCIgD+CD40L+101 in comparison
with the pCIgD group was observed and, also, interferon gamma, interleukin (IL)-6, and IL-17A levels were
higher. Considering the results obtained from this study for humoral and cellular responses in mice, the
inclusion of pCD40L and 101 as adjuvants in a BoHV-1 DNA vaccine for cattle is highly recommendable.

Keywords: BoHV-1 DNA vaccine, CD40L, chemical adjuvants, cytotoxic response, mice model

Introduction abortion. More importantly, infected animals that are im-


munosuppressed may suffer secondary bacterial infections

B ovine herpesvirus-1 (BoHV-1) is an important viral


pathogen of cattle, and it produces a variety of diseases,
including vulvovaginitis/balanoposthitis, infectious bovine
increasing morbidity and mortality (21,36).
However, if infection or reactivation of the virus occur
during pregnancy, abortions can occur (54,59,70). Six Eu-
rhinotracheitis, systemic infection in the newborn, and ropean countries became BoHV-1 free after the introduction
abortion in cows (35,73). The economic impact of this in- of expensive eradication programs, consisting of seroposi-
fection related to weight loss, decreased milk production, and tive animals culling (62,71).

1
Instituto de Virologı́a-IVIT (INTA-CONICET), Buenos Aires, Argentina.
2
Universidad Nacional de Rio Negro, Sede Atlántica, Viedma, Rı́o Negro, Argentina.
3
Laboratorio de células presentadoras de antı́geno y respuesta inflamatoria. Instituto de Medicina Experimental (IMEX) - CONICET,
Academia Nacional de Medicina, Buenos Aires, Argentina.
4
Cátedra de Inmunologı́a Aplicada, Universidad del Salvador, Buenos Aires, Argentina.
*These authors contributed equally to the results presented in this report.

1
2 LANGELLOTTI ET AL.

A more common way to prevent BoHV-1-related dis- secreted form of BoHV-1 gD, in sheep, enhanced the humoral
eases is the use of inactivated or attenuated live vaccines. immune response to a plasmid coding for gD alone (47). On
However, they cannot control latent infection. The ability to the contrary, immunized calves with the same vaccine had
differentiate infected from vaccinated animals (DIVA) is an significantly higher gD-specific serum titers of immuno-
important issue (65). Live-attenuated vaccines may cause globulins G and A than plasmid gD vaccine alone, although
abortion, latency with reactivation and shedding (79,80). protection upon BoHV-1 challenge was not achieved (48).
Also, they may downregulate major histocompatibility Mice have been used as an experimental model to select
complex (MHC) class I molecules (31,55), which com- optimal formulations for BoHV-1 vaccines before carrying
promise the subsequent cellular immune response. On the out experiments in large animal hosts (67).
contrary, inactivated viral vaccines have a short duration of We have previously shown in the mouse model that in-
immunity and are poor inducers of cellular responses (17). corporation of adjuvants into a genetic vaccine against
Importantly, BoHV-1 can persist in the presence of antiviral- BoHV-1 is immunologically beneficial (18,40). Recently, we
specific antibodies (24,38,52). So, cytotoxic T lymphocytes demonstrated that bovines vaccinated with a DNA vaccine
(CTLs) are necessary for controlling the virus (73). Novel coding BoHV-1 gD, adjuvanted with Montanide 1113101PR
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

vaccines against BoHV-1 that do not have the problems as- (101), induced better protection upon challenge than animals
sociated with current vaccines are thus necessary. They need vaccinated with a genic vaccine alone. Although this vaccine
to contain viral epitopes capable of inducing protective im- was able to diminish clinical symptoms and viral excretion,
mune responses, and should ideally be suitable for DIVA. our results showed that it is still necessary to induce a higher
DNA vaccines have potential to induce cell-mediated and immune response against the virus (61).The 101 adjuvant is
humoral immune responses with the advantages of having composed of non-crosslinked charged polymers (10–500 nm)
good thermostability, the capability of coexpression of dispersed in water with an immunostimulating compound. It
multiple antigens and reduced production costs since they has the capacity to improve DNA binding and transfection
avoid the need of working with viral particles (4,10,28). potency. Also, it can recruit cells to the site of injection (78).
Licensed DNA vaccines exist for hematopoietic necrosis In this study, we evaluate the immune response induced
virus in salmon (Apex-IHN, Novartis) (26), West Nile virus in mice by a DNA vaccine containing the truncated, secreted
in horses (West Nile Innovator, Fort Dodge) (15), and version of BoHV-1 gD (18), in combination with the adju-
melanoma in dogs (Canine Melanoma Vaccine, Merial) (6). vants CD40L and 101, to extend our previous studies in the
Although genic vaccines have the capacity to induce murine model.
cytotoxic responses (40), they have limited potency because
they do not amplify or propagate ‘‘in vivo.’’ For this reason, Materials and Methods
it is necessary to increase the magnitude and direction of the
Plasmid construction
immune response with the incorporation of molecular and
chemical adjuvants. Polymerase chain reaction (PCR) product encoding the
Viral glycoproteins are involved in several steps of secreted version of the BoHV-1 glycoprotein D (gD) was
BoHV-1 replication (7,25,43,57). Glycoprotein D (gD) is inserted into EcoRI-digested pCIneo vector (Promega) to
responsible for the penetration of the BoHV-1 in cells with construct plasmid pCIgD, prepared as described before (40).
participation in the membrane fusion and viral adsorption Plasmid pDNA3.1 contained the gene for murine CD40L
(74). It induces neutralizing antibodies (3,13,17) and has (pCD40L) and was kindly provided by Dr. Kipps, University
cytotoxic epitopes (16,17). Notably, vaccination with a of California. Nonrecombinant plasmid pCIneo was used as
plasmid encoding the secreted form of the gD encoding in a control. All plasmid preparations had A260/280 ratios >1.7.
vector produced a different immune response (type 1/type 2) After purification, plasmids were diluted with phosphate-
than vaccination with one encoding full-length gD (41,42). buffered saline (PBS).
Injection of a naked plasmid vector into animal muscle or
skin can induce an immune response against a transgene- Adjuvant
encoded antigen (Ag) (60), but in general this response is
Montanide 1113101PR (101) was provided by Seppic
weak and needs to be enhanced to provide protective im-
Inc., France, and used according to the manufacturer’s
munity (23,49,61). The interaction between the CD40 re-
instructions.
ceptor and its ligand, CD40L, regulates humoral and cellular
Toxicological tests made on Montanide adjuvants (derml
immune responses. CD40L (or CD154) is a 39-kDa type II
irritation test, Oral LD 50, Berlin test, IP LD 50, pyroge-
membrane glycoprotein with homology to TNF-a (2). This
nicity, ocular irritation test) can be considered nontoxic and
molecule is expressed transiently on CD4+ T cells within
well tolerated. The Committee for Veterinary Medical
hours after ligation of the TCCRD 3 complex and then is
Products (CVMP) considered these adjuvants safe for use in
rapidly downmodulated (66). CD40L allows the T lympho-
immunological products. These substances have been au-
cyte to interact with B lymphocytes, monocytes, and den-
thorized in the annex of the European Council Regulation n
dritic cells (DCs), inducing their proliferation, differentiation,
470/2009 (previously 2377/90/EC).
and expressing accessory molecules that facilitate the pre-
sentation of antigens to T lymphocytes (22). Given the ability
Generation of bone marrow-derived DCs
of CD40L to enhance the development and proficiency of
antigen-presenting cells (APCs), we hypothesized that coin- Bone marrow-derived DCs were obtained as previously
jection of a DNA vaccine with a plasmid encoding CD40L described (39). In brief, the marrows of femurs and tibiae of
could improve the immune response to the transgene antigen. naı̈ve BALB/c mice (n = 3) were flushed out with RPMI
Inoculation of a plasmid coding for bovine CD40L fused to a 1640 medium. After washing, cells were diluted at 1 · 106
CYTOTOXIC ACTIVITY USING A DNA VACCINE 3

cells/mL in RPMI 1640 medium (Invitrogen Life Technol- reverse primers: 5¢-AAGAATTCATGTTGCCTACACCCG
ogies) with 10% fetal calf serum (FCS), 5.5 · 10 - 5 of 2- CGCCGCGGGT-3¢ and 5¢-AAGAATTCTCAGGCGTCGG
mercaptoethanol (Sigma-Aldrich), and 30% conditioned GGGCCGCGGGCGTA-3¢ that contain EcoRI restriction
medium from GM-CSF-producing NIH-3T3 cells. The cells sites and amplify a 1005 bp fragment (nt 138 to 1143) of the
were cultured for 9 days with 50% of the medium replaced gD gene (emb AJ004801 BHV1CGEN). The ATG start codon
every 2 days. On day 9 of culture, >85% of the harvested for expression in bacteria is provided by the pRSETB vector
cells expressed CD40, CD11c, MHC class II, and CD80 but before a 6 His-codon stretch, while the reverse primer includes
no Gr-1 (data not shown). the gene stop codon. The 1005 bp fragment was inserted into
plasmid pRSET-B (Invitrogen) to produce pRSETgD. The
Endocytosis assay correct orientation and the integrity of the gD sequence were
confirmed by nucleotide sequencing and restriction analysis
DCs (produced from naı̈ve mice) were incubated with
(data not shown). E. coli BL21 cells were used to express the
medium, pCIneo (1 lg/mL), pCIgD (1 lg/mL), 101 adjuvant
recombinant plasmid by standard methods. Expression was
(ratio 24% adjuvant and 76% PBS), and pCIgD+101 (ratio
induced with 1 mM IPTG (Sigma) for 2 h when the absor-
24% adjuvant and 76% DNA). Then, 100 lg/mL FITC-
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

bance was 0.3 at 600 nm. Proteins were extracted with 6 M


labeled OVA was added to RPMI 1640 medium and incu-
guanidine HCl and 0.5 M NaCl (denaturing buffer), and gD-
bated for 40 min at 37C. After incubation, the cells were
his protein was purified by Ni-nitrilotriacetic acid (Ni-NTA)
washed with cold PBS three times, and evaluation of uptake
Agarose columns (Invitrogen).
was tested by flow cytometry. The fluorescence background
in cells incubated with FITC-OVA at 4C was determined
and subtracted. The results are expressed as mean fluores- ELISA for detection of anti-BoHV-1 antibodies
cence intensity values. Immulon 1 plates were coated with 50 lL of inactivated
BoHV-1 (iBoHV-1) (2 · 107 TCID50/mL) in 0.1 M
Animal treatment and vaccine formulations carbonate-bicarbonate buffer, pH 9.6. The plates were incu-
Male BALB/c mice (28 – 2 g) from La Plata University bated overnight (ON) at 4C. They were blocked, and serial
(Argentina) were used. The animals were acclimated and dilutions (1:10) of mice sera were diluted in PBS/0.05%
randomly distributed into five experimental groups. Mice Tween 20 (PBST) containing 1% ovalbumin (PBST-OVA)
were kept in rodent caging with ventilated systems, and re- and dispensed in 50 lL/well aliquots. After incubation, plates
ceived water and food ad libitum. Handling and housing of were washed with PBST, and antimouse IgG peroxidase
animals were according to the guidances of the Institutional conjugate (KPL) was added for 1 h at 37C. Finally, ortho-
Committee for Care and Use of Experimental Animals, phenylene-diamine (1,2-benzenediamine) dihydrochloride
CICUAE-INTA, Argentina (Approved Protocol n 54/2016). (OPD; SIGMA) and H2O2 were added, and absorbance was
Groups of 5 mice were intradermically (i.d.) inoculated in measured at 492 nm in a MR 5000 microplate reader (Lab-
the back, at days 0 and 15, with 15 lg of pCIgD and 75 lg systems, MN). The cutoff was established as the mean A492
of pDNA3.1-mCD40L (pCD40L) formulated without and of the negative sera +2 standard deviations. Titers were ex-
with Montanide 101 adjuvant in a final volume of 0.2 mL pressed as the log10 of the reciprocal of the highest serum
(distributed in two sites) (The dose of plasmid injected dilution giving an A492 higher than the cutoff.
was previously selected based on dose-response curve). The
same amount of pCIneo was inoculated as negative control IgG1, IgG2a, IgG2b, and IgG3 isotyping ELISA
group. Serum samples were obtained at days 14 and 30. Greiner microtiter plates were coated with purified re-
combinant gD (1,4 lg/well) to evaluate the IgG1, IgG2a,
Virus IgG2b, and IgG3 subtypes in vaccinated mice. Plates were
BoHV-1 strain LA (Los Angeles) was propagated in blocked with PBST containing 0.5% gelatine (PBST-G), and
Madin Darby bovine kidney (MDBK) cells, in E-MEM then serial dilutions (1:10) of mice sera were prepared in
medium supplemented with 10% fetal bovine serum (Gib- PBST-G and added to plates in 50 lL/well aliquots, for 2 h
co). The virus was added at a multiplicity of infection (moi) at room temperature. PBST was added to wash the plates.
of 0.1, and it was allowed to adsorb for 45 min at 37C Biotinylated goat antimouse IgG1, IgG2a, IgG2b, and IgG3
before the addition of more culture medium. The superna- (Caltag Laboratories, San Francisco, CA) were dispensed
tant was collected, and cell debris was removed by centri- and after 60 min incubation, plates were washed, and anti-
fugation at 23,699 g for 1 h at 4C, when cytopathogenic mouse IgG peroxidase conjugate (KPL) was added and in-
effects were observed. Supernatants of infected cells were cubated for 1 h at room temperature. The reaction was
pelleted by ultracentrifugation at 120,000 g for 1 h at 4C. visualized as described in section ‘‘ELISA for detection of
The virus suspension was stored at -70C. Inactivated virus anti-BoHV-1 antibodies.’’
(30 min at 11 cm from two General Electric G875 ultraviolet
bulbs) was used for enzyme-linked immunosorbent assay Percentage of BoHV-1 neutralization
(ELISA) and in vitro cell stimulation.
When seroneutralizing antibodies were measured in sera
of immunized mice at 30 dpv using 100 DICT50 infective
Assembly of gD expression vector and production
tissue culture doses, all values were negative. So, it was
of recombinant gD
decided to measure the percentage of neutralized virus in-
The sequence coding for full-length gD was amplified by cubated 1/8 serum dilution at 25 lL of volume with different
PCR from BoHV-1 DNA, using the following forward and fivefold dilutions of infective BoHV-1 (1000 to 1 TCID50
4 LANGELLOTTI ET AL.

per 25 lL) in quadruplicates. Then, the virus–serum mix- In vitro CFSE proliferation assay
ture, previously incubated ON at 37C under 5% CO2, was Carboxylfluorescein diacetate succinimidyl ester (CFSE;
seeded on MDBK cell monolayers. After 1 h at 37C, cells 5 nM) (BD Biosciences) was used to label splenocytes
were washed with PBS. Finally, cells were incubated with for 40 min at 37C. Cells were washed four times and re-
fresh medium supplemented with 2% FCS at 37C under 5% suspended in RPMI complete medium. The cells were cul-
CO2. After 72 h, cytopathic effects were observed. The tured with medium, iBoHV-1 or Concanavalin A for 4 days.
remnant infective virus was determined by a TCID50 assay. Then cold PBS was used to wash the splenocytes. Fixation
The calculated viral titer was expressed as the percentage of was done by resuspension in 0.2% paraformaldehyde in
BoHV-1 that was still infective after serum incubation, FACSFlow (BD, Buenos Aires, Argentina) and analyzed
compared with the titer of virus suspensions incubated by cytometry.
without serum (a 25 lL volume of medium was incubated
with the different dilutions of infective BoHV-1).
Statistical analysis
Cytokines detection Data between three or more groups were analyzed using
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

one-way analysis of variance (ANOVA) and Dunnett’s post-


Spleens were extracted from mice, and splenocytes were
tests. Differences were considered significant at p-value <0.05.
disaggregated by gentle homogenization in RPMI 1640
medium/10% FCS (Gibco)/1% ampicillin-streptomycin, 15
days after the booster (30 dpv). In 96-well flat-bottomed Results
plates, pool of splenocytes from each group were plated pCIgD plus 101 adjuvant enhances
(1 · 106 cells per well) in 100 lL complete medium [RPMI endocytosis effect on DCs
1640/1% ampicillin-streptomycin/1 mM sodium pyruvate
(Gibco)/10% FCS/5 mM HEPES (Gibco)/2 mM L-glutamine Immature DCs are specialized to sense the microenviron-
(SIGMA)/50 mM 2-mercaptoethanol/10 mM MEM Non- ment and incorporate the antigen through phagocytosis or
essential Amino Acids Solution (Gibco)]. In triplicate 100 lL endocytosis. With the aim to determine if the selected adju-
of inactivated BoHV-1 was added to the wells in complete vant for the genic vaccine or the DNA vaccine were able to
medium or medium alone, and incubated for 48 h at 37C. affect the antigen uptake by endocytosis in naı̈ve DCs, these
The supernatant was collected, and interferon gamma (IFNc), cells were cultured in vitro with pCIneo, pCIgD, pCIgD+101,
interleukin (IL)-4, IL-6, IL-17A, and IL-10 were measured 101 adjuvant alone, or medium like a basal control. Finally,
using BD CBA Kits (Mouse Th1/Th2/Th17) by flow cy- cells were incubated with OVA coupled to FITC for 40 min at
tometry. The results were expressed in pg/mL. 37C. Since this is a temperature-dependent phenomenon, the
reaction control was DCs incubated with OVA-FITC at 4C.
Endocytosis levels were then measured by flow cytometry.
Cytotoxic T cell assay ( JAM test)
The 101 adjuvant plus pCIgD produced a significant in-
Cytotoxicity assay was previously described by Matzin- crease of endocytosis in these cells. On the contrary, pCIgD,
ger et al. (50). In brief, splenocytes were isolated as de- pCIneo, or 101 adjuvant alone did not modify endocytosis
scribed in section ‘‘Cytokines detection.’’ Cells were levels (Fig. 1).
cultured with inactivated BoHV-1 or medium alone for 5
days at 37C and 5% CO2. P815 cells infected with a moi of
5 of BoHV-1 and labeled with [methyl-3H] thymidine were
used as target cells. 104 P815 cells per well were plated with
dilutions of effector cells (splenocytes) and incubated in
triplicate for 3 h at 37C. The effector/target ratio used in the
assay was 80:1. A semiautomatic cell harvester (Skatron)
was used to harvest the cells. Radioactivity was measured
by liquid scintillation counting (experimental release).
Spontaneous release was determined in target cells incu-
bated with complete medium only. Percentages of specific
lysis were calculated using the formula:
% Specific lysis = 100 · [(spontaneous release – experi-
mental release)/spontaneous release].

Immunofluorescent staining
The following labeled monoclonal antibodies were used
to stain the surface markers of splenocytes: allophycocyanin
anti-CD8b, fluorescein isothiocyanate antimouse CD4, and
conjugated rat Ig isotype controls (Pharmingen). Spleno-
FIG. 1. Adjuvant effect on BMDC. Endocytosis of OVA-
cytes were incubated with labeled antibodies for 20 min at FITC antigen by BMDC cultured in vitro with different
4C. Then, they were washed with PBS containing 1% FCS formulations for 48 h was tested. Results are expressed as
and fixed with 0.2% paraformaldehyde. The stained cells the MFI – SEM of five independent experiments. BMDC,
were evaluated by flow cytometry in a BD FacsCalibur and bone marrow-derived dendritic cells; MFI, mean fluores-
analyzed with CellQuest software. cence intensity; SEM, standard error of the mean. *p < 0.05.
CYTOTOXIC ACTIVITY USING A DNA VACCINE 5

In previous studies, cattle were immunized with pCIgD by cytometric bead arrays on a pool of supernatant of
and adjuvant 101. After viral challenge, higher protection splenocytes stimulated or not with inactivated BoHV-1 or
levels were observed as compared with animals immunized concanavalin A, as a positive control, from each group of
with pCIgD alone (61). For this reason, adjuvant 101 was vaccinated mice.
selected for this study, with the addition of a plasmid en- The pCIgD vaccine group secreted lower levels of IFNc,
coding CD40L, to enhance the immune response against gD. IL-4, IL-6, IL-17, and IL-10 cytokines than the other for-
mulations containing pCIgD (Fig. 3A–E).
pCIgD vaccines formulated with pCD40L, 101 The amount of IFN-c produced upon vaccination with
adjuvant, and pCD40L plus 101 elicit higher specific pCIgD+pCD40L (694.64 pg/mL) was *20 times higher
humoral immune responses than a pCIgD vaccine. than the amount produced upon vaccination with pCIgD
Neutralizing capacity of the sera alone (32.49 pg/mL), and 11 times higher than those in the
pCIgD+101 or pCIgD+pCD40L+101 groups (Fig. 3A).
Groups of mice were vaccinated with 15 lg of pCIgD (per
Also, in the case of IL-6 and IL-10, pCIgD+pCD40L group
dose) formulated with and without pCD40L, 101 adjuvant,
is the major producer of both cytokines as compared with
and a combination of both, and the elicited immune re-
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

the other groups (Fig. 3B, C). To highlight in this sense


sponses were evaluated and compared. Analysis of sera by
that the pCIgD+pCD40L+101 group secrete similar levels
ELISA to BoHV-1 showed that at 14 dpv, all animals in
of IL-6 than the pCIgD+pCD40L group. In case of IL10,
groups immunized with pCIgD+pCD40L, pCIgD+101, and
the pCIgD+pCD40L group was the only group that pro-
pCIgD+pCD40L+101, were able to induce specific BoHV-1
duces this cytokine (Fig. 3C).
antibodies. Moreover, at 30 dpv, levels of antibodies against
Besides, low levels of IL-4 were detected among the
BoHV-1 in groups pCIgD+pCD40L, pCIgD+101, and
pCIgD, pCIgD+pCD40L, and pCIgD+pCD40L+101 groups,
pCIgD+pCD40L+101 were significantly higher than those in
pCIgD+101 being the major producer group of this cytokine
the pCIgD group ( p < 0.01). None of the pCIneo-immunized
(Fig. 3D).
control animals produced any specific immune response at
Interestingly, production of IL-17A could be detected in
any time compared with normal mouse serum (Fig. 2A).
the pCIgD+pCD40L, pCIgD+101, and pCIgD+pCD40L+101
Moreover, analysis of sera by ELISA showed that specific
groups but not in the pCIgD one (Fig. 3E). Animals im-
antibodies against the gD-protein were induced at 14 dpv.
munized with pCIneo only secreted low levels of IL-6.
Total antibody levels were significantly increased ( p < 0.05)
at 14 dpv in the pCIgD+101 group compared with the pCIgD
Combination of pCIgD with pCD40L and 101 adjuvant
group. In contrast, at 30 dpv all immunized groups showed
increase splenic CD4 + T lymphocytes cell population
the same high antibody titers (Fig. 2B).
Increases in IgG isotype induced by the different vaccines Spleen cells obtained from vaccinated mice were used to
were studied next. At 14 dpv, IgG2a and IgG2b were the main determine the effect of these vaccines on T lymphocyte
isotypes in the sera of mice vaccinated with pCIgD+CD40L, populations.
and were significantly increased with respect to the pCIgD A significant ( p < 0.05) expansion of CD4+ T cells popu-
group (IgG2a p < 0.05; IgG2b p < 0.01). The same was ob- lation was observed at 30 dpv in the pCIgD+101+pCD40L
served in the group immunized with pCIgD+101 adjuvant group as compared with pCIneo and pCIgD (Fig. 4A). No
(vs. pCIgD: IgG2a, p < 0.05; IgG2b, p < 0.001) and with significant differences, on the contrary, were observed in the
pCIgD+pCD40L+101 adjuvant (vs. pCIgD: IgG2a, p < 0.01; CD8+ lymphocyte populations measured (Fig. 4B). In the other
IgG2b, p < 0.001). Furthermore, group pCIgD+101 presented groups, no changes were observed in these two populations.
significantly higher levels of specific IgG1 ( p < 0.05) than the
pCIgD group. At 30 dpv, although specific gD isotype titers Vaccination with pCD40L +101 enhances the specific
were high, IgG1 and IgG3 showed higher levels, and no sig- cytotoxic T cell in vivo response against BoHV-1
nificant differences between groups were observed (Fig. 2C).
Using a specific viral cytotoxicity test ( JAM test), sple-
The viral neutralizing capacity of sera from immunized
nocytes of pCIgD mice with or without pCD40L and/or 101
animals was studied in a 1/8 dilution. At 30 dpv (Fig. 2D), the
adjuvant and pCIneo groups were restimulated in vitro with
average percentages of neutralized virus were 1.8 for sera in
UV-inactivated BoHV-1 and then incubated with radioac-
the pCIneo group, 18.4, 35.7, 55.7, and 24.4 for the pCIgD,
tively labeled BoHV-1-infected P815 cells.
pCIgD+CD40L, pCIgD+101, and pCIgD+CD40L+101 group,
CTL activities in the three groups that received pCIgD
respectively. However, when antibodies were evaluated in a
plus pCD40L and/or 101 adjuvant, which were restimulated
neutralization test with a fixed amount of virus, no serum
in vitro with iBoHV-1, were significantly higher
sample at 30 dpv was able to neutralize 100 TCID50 of
(pCIgD +pCD40L, 21.12% p < 0.01; pCIgD+101, 18.04%
BoHV-1 (virus neutralization titers <0.9, data not shown). This
p < 0.05; pCIgD+pCD40L+101, 26.22% p < 0.001) than those
assay evidenced a weak capacity of the antibodies to neutralize
in the pCIgD group (6,95%) (Fig. 5). However, when a cutoff
virus in all animals. However, a differential neutralizing capacity
was selected (pCIneo mean – 2DS = 20% specific cytotoxicity)
was observed among groups, being the animals immunized with
only pCIgD+pCD40L+101 had all animals over the cutoff with
pCIgD+101 those that managed to neutralize the largest amount
specific positive cytotoxicity activity. In contrast, only one and
of viral particles in comparison to the other groups.
two animals were positive in the pCIgD+pCD40L and the
pCIgD+101 groups, respectively. These results indicate that the
Cytokine production
gD-encoding plasmid plus pCD40L, 101 adjuvant, or both
To profile the cytokine levels induced by different vac- induce specific CTL that recognizes BoHV-1-infected cells.
cines at 30 dpv, production of some cytokines was evaluated Yet, only combined with 101 and pCD40L, pCIgD has the
6 LANGELLOTTI ET AL.
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

FIG. 2. Humoral immune response elicited. (A) Humoral response in mice vaccinated with different formulations. Mice
were intradermally immunized on days 0 and 15, and serum IgG antibody levels specific for BoHV-1 were measured at 14
and 30 dpv. Bars represent the mean of log10 antibody titers from mice in each group – SEM. Significant differences with
respect to the pCIgD group are indicated as **( p < 0.01). (B) gD-specific antibodies detected by ELISA at 14 and 30 dpv.
The results are presented as the mean – SEM of log10 antibody titers. Significance of differences with respect to the pCIgD
group are indicated as *p < 0.05. (C) Anti-gD-specific immunoglobulin isotypes measured by indirect ELISA at 14 and 30
dpv. Data represent the mean – SEM of log10 antibody titers. Significant differences from the mean of the pCIgD group are
indicated as *p < 0.05, **p < 0.01, and ***p < 0.001. (D) Neutralization of BoHV-1 by sera from immunized animals. A 1:8
serum dilution from each group (pCIneo, pCIgD, pCIgD+101, pCIgD+CD40L, and pCIgD+CD40L+101) was incubated with
different BoHV-1 doses, and the remnant infective virus was determined by a TCID50 assay. Titer reductions are expressed
as % neutralized BoHV-1 (black bars), and the remnant infective virus is expressed as % non-neutralized BoHV-1 (white
bars). BoHV, bovine herpesvirus-1; ELISA, enzyme-linked immunosorbent assay.

capability to achieve consistently high specific cytotoxicity Discussion


toward BoHV-1 in all animals of the group. The major problem to control BoHV-1 resides in the ca-
pability of the virus to produce latency and immune evasion
in infected animals. During infection, the virus propagates
Vaccination with pCIgD +pCD40L or 101 adjuvant
intracellularly followed by an extracellular dispersion, and
enhances BoHV-1 specific T cell proliferation
persists even in the presence of neutralizing antibodies. Thus,
Since the cellular immune response is essential to control a BoHV-1 vaccines should induce humoral and cellular im-
BoHV-1 infection, specific T cell proliferative responses mune responses with induction of CTL (44,46).
were measured. As shown in Table 1, there are more animals Huang et al. (32) used a plasmid coding BoHV-1 gly-
with a viral-specific cell proliferative response considered as coprotein B (gB) to induce cellular and humoral immune
positive (above the cutoff = 3.64) in the pCIgD+101 and responses in mice, and showed production of specific anti-
pCIgD+pCD40L groups (60% positive proliferation in both bodies and CTL responses. The same vaccine induced hu-
groups against 25% in the pCIgD group). Surprisingly, under moral responses and elicited IFNc secreting CTL in calves
these experimental conditions, no specific proliferation was (32). This was the first demonstration that the plasmid in-
detected in the pCIgD+pCD40L+101 group. All splenocytes duced CTL not only in mice but also in calves. The most
stimulated with concanavalin A had a proliferative response. important glycoprotein in BoHV-1 is glycoprotein D (gD),
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

FIG. 3. Cytokine detection with CBA. Splenocytes were stimulated or not for 48 h with inactivated BoHV-1 or conca-
navalin A, as a positive control, and cytokine production was detected using a Cytometric Bead Array (CBA) kit. (A) IFNc,
(B) IL-6, (C) IL-10, (D) IL-4, (E) IL-7A. Each bar represents a pool of supernatants of splenocytes from each mouse group.
IFNc, interferon gamma; IL, interleukin.

FIG. 4. Cellular spleen subpopulations from vaccinated mice at 30 dpv. Each point corresponds to one animal of the
group. Results are expressed as population percentages (means – SEM) (A) CD4 lymphocytes (CD4+); (B) CD8 lympho-
cytes (CD8b+). *p < 0.05.

7
8 LANGELLOTTI ET AL.
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

FIG. 5. Immunization with pCIgD and adjuvants induces an enhanced CTL response as compared with pCIgD alone.
Splenocytes collected at 30 dpv and restimulated in vitro with BoHV-1, and were tested for CTL response by a cytotoxicity
assay against the virus. Representative results of percentage specific lysis are shown at an effector/target ratio of 80:1.
*p < 0.05, **p < 0.01, and *** p < 0.001 (statistical significance of differences with respect to the pCIgD group). A cutoff of
20 was calculated as the mean of the pCIneo group +2 SD. CTL, cytotoxic T lymphocytes.

which is involved in the protective immune response (3,33),


with major targeting to CD8+ CTL (16,17,40), and with the
production of neutralizing antibodies (3,17,33,58,76,77,82).
Table 1. T Cell Proliferation in Response In this study, a secreted form of BoHV-1 gD was chosen
to Vaccination with pClgD Alone because previous studies of others performed in mice and
and Formulated with 101 and/or CD40L cattle showed that this truncated version of gD induced
higher antibody titers than the membrane-anchored form
Treatment N animal % Proliferation* (41,42,75).
Given the importance of adjuvants to increase vaccine
pClneo 1 0.56 efficiency, the immunomodulatory effects on a DNA vac-
2 1.78
3 2.94 cine of Montanide 1113101PR (101) adjuvant (SEPPIC)
4 1.38 were previously analyzed in our laboratory. We demon-
pClgD 1 3.02 strated that a DNA vaccine based on BoHV-1 gD formu-
2 0.16 lated with 101 adjuvant induced not only specific antibodies
3 3.52 but also cellular immunity against BoHV-1 with viral-
4 4.35a specific T cell proliferation and the high levels of IFNc
pClgD+pCD40L 1 6.59 secretion in cattle (33).
2 5.80 This study examined the benefits of the CD40 ligand,
3 2.83 CD40L. This molecule belongs to the tumor necrosis
4 2.01 factor superfamily, hasa transient expression in activated
5 3.75
pClgD+101 1 6.51 T lymphocytes, and can stimulate DCs and B cells (27).
2 3.79 This stimulation activates DCs and primes the CD8+
3 1.31 T cell response (5,64,68). Also, CD40L increases the
4 3.96 survival and differentiation of activated B cells (20).
5 0.60 These qualities make CD40L an attractive vaccine adju-
pClgD+pCD40L+101 1 0.10 vant (8,30,34,51,69,81).
2 0.97 Incorporation of any of these adjuvants (101, pCD40L,
3 2.52 or both) to gD vaccine formulations yielded an increased
4 1.64 humoral response toward BoHV-1 and gD as well. Similar
5 0.51 results for 101 and CD40L were reported by Quattrocchi
Spleen cells were isolated from animals inoculated with pCIgD et al. (61) and Manoj et al. (48), respectively. Vaccines
with adjuvants and in vitro stimulated with iBoHV-1 for 4 days, formulated with these adjuvants yielded high levels of
followed by measurement of T cell proliferation by lost of CFSE. antibodies in cattle, but insufficient protection upon
* % Proliferation = % proliferation stimulated with BoHV-1 - % BoHV-1 challenge.
proliferation stimulated with medium.
a
Results in bold and underline are over the cutoff. The cutoff is Analysis of isotypes in serum showed that IgG1, IgG2a,
calculated as pClneo mean + 2 SD (cutoff = 3.64). IgG2b, and IgG3 levels were similar in all animals, but the
BoHV, bovine herpesvirus-1; SD, standard deviation. major variation was observed in IgG2a and IgG2b at 14 dpv.
CYTOTOXIC ACTIVITY USING A DNA VACCINE 9

An increase in IgG2a has been described for DNA vaccines Acknowledgments


(63) and suggests a T-helper 1 response. Predominance of The authors thank Dr. Osvaldo Zabal for the valuable
IgG2a has been described for humoral protective immune assistance with cell cultures, Seppic for providing the
responses to virus in mice. In these animals, this isotype is Montanide ESSAI 1113101PR adjuvant, Dr. Kipps for pro-
clearly more efficient at complement fixation (11,12,37,56) viding the pDNA3.1-mCD40L in this study, and Ms. Pamela
and as opsonizing agent than IgG1 (1). Detection of lym- Angeletti and Fátima Torales for their technical assistance.
phocyte proliferation after inactivated BoHV-1 stimulation
and isotype switch evidenced by IgG2a secretion indicates a
balance to a Th1/Th2 profile of CD4+ T cells (11,12,53,72). Author Disclosure Statement
One feature of adjuvants is their inflammatory effect that No competing financial interests exist.
attracts inflammatory cells, including macrophages and DCs
(14,78). This characteristic plays an important role in the
immune response. On the contrary, the formulation of Funding Information
pCIgD with 101 adjuvant can be efficiently endocytozed by This work was supported by INTA Project PNBIO
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

APCs, generating an increase in the proliferative response of 1131032 and by a grant from Agencia Nacional de Pro-
other cells (29). It can be hypothesized that in the present moción Cientı́fica y Tecnológica PICT2014-0891.
experiments the 101 adjuvant recruited APCs at the site of
inoculation, facilitating DNA internalization, as observed by
References
Dupuis and collaborators (19).
Since cellular immune response is important to deal with 1. Abbas AK, Murphy KM, and Sher A. Functional diversity
BoHV-1 infection, the specific T cell proliferative response of helper T lymphocytes. Nature 1996;383:787–793.
was measured. Our results showed that this response was 2. Armitage RJ, Fanslow WC, Strockbine L, et al. Molecular
incremented in the pCIgD+101 and pCIgD+pCD40L groups and biological characterization of a murine ligand for
(60% positive proliferation in both groups against 25% in CD40. Nature 1992;357:80–82.
the pCIgD group). Surprisingly, under these experimental 3. Babiuk LA, ’Italien JL, van Drunen Littel-van den Hurk S,
conditions no specific proliferation was detected in et al. Protection of cattle from bovine herpesvirus type I
pCIgD+pCD40L+101 group. This could be due to expen- (BHV-1) infection by immunization with individual viral
sive cell death after 4 days of proliferation, when cells glycoproteins. Virology 1987;159:57–66.
were harvested. 4. Babiuk LA, van Drunen Littel-van den Hurk S, Loehr BI,
et al. Veterinary applications of DNA vaccines. Dev Biol
Genetic immunizations were used in several animal
(Basel) 2000;104:73–81.
models and hosts, such as mice and cattle (9,13,61,75). It
5. Bennett SR, Carbone FR, Karamalis F, et al. Help for
has been demonstrated by Deshpande et al. (17) that in- cytotoxic-T-cell responses is mediated by CD40 signalling.
duction of specific CTL against BoHV-1 was elicited by Nature 1998;393:478–480.
membrane-anchored gD. The same phenomenon was 6. Bergman PJ, Camps-Palau MA, McKnight JA, et al.
demonstrated by our group inoculating a DNA vaccine Development of a xenogeneic DNA vaccine program for
encoding a secreted version of gD (40). The pCIgD vac- canine malignant melanoma at the Animal Medical Center.
cine was formulated with 101 and pCD40L with the ex- Vaccine 2006;24:4582–4585.
pectation of inducing CTL against the virus, and a specific 7. Brum MC, Coats C, Sangena RB, et al. Bovine herpesvirus
cytotoxicity assay was set up with BoHV-1-infected P815 type 1 (BoHV-1) anterograde neuronal transport from tri-
cells. This experiment demonstrated that the mice vacci- geminal ganglia to nose and eye requires glycoprotein E.
nated with different formulations had the capacity to elicit J Neurovirol 2009;15:196–201.
CTL production. We observed the occurrence of memory 8. Burger JA, Mendoza RB, and Kipps TJ. Plasmids encoding
CD8+ cells that recognized and lysed P815-infected cells. granulocyte-macrophage colony-stimulating factor and
The specific cytotoxicity was consistent with the increases CD154 enhance the immune response to genetic vaccines.
in IL-17A detected in pCIgD+101, pCIgD+pCD40L, and Vaccine 2001;19:2181–2189.
pCIgD+101+pCD40L. IL-17A is required for optimal 9. Caselli E, Boni M, and Di Luca D. A combined bovine her-
specific CD8+ T cells activity. In addition to Th17 cells, pesvirus 1 gB-gD DNA vaccine induces immune response in
there are other sources of IL-17, such as Tcd, NKT, and mice. Comp Immunol Microbiol Infect Dis 2005;28:155–166.
CD8+ T cells (45,83). In DCs, IL-17A upregulates surface 10. Cichutek K. DNA vaccines: development, standardization
MHC class I molecules and enhances production of IL-6. and regulation. Intervirology 2000;43:331–338.
11. Coutelier JP, van der Logt JT, and Heessen FW. IgG subclass
This facilitates crosspriming and proliferation of CTL. In
distribution of primary and secondary immune responses con-
the same sense, secretion of IFNc indicates the activation
comitant with viral infection. J Immunol 1991;147:1383–1386.
of a Th1-type response. 12. Coutelier JP, van der Logt JT, Heessen FW, et al. Virally
Taking together, our results indicate that formulation of a induced modulation of murine IgG antibody subclasses.
pCIgD DNA vaccine with adjuvants 101 and/or pCD40L J Exp Med 1988;168:2373–2378.
elicits a higher humoral and cellular immune response 13. Cox GJ, Zamb TJ, and Babiuk LA. Bovine herpesvirus 1:
against this virus than the plasmid alone in mice. They also immune responses in mice and cattle injected with plasmid
show that only when both adjuvants are included, CTL is DNA. J Virol 1993;67:5664–5667.
increased in all immunized animals. Because of this, we 14. Cox JC, and Coulter AR. Adjuvants—a classification and
suggest that a pCIgD vaccine formulated with 101 and review of their modes of action. Vaccine 1997;15:248–256.
pCD40L should be tested in bovines since this formulation 15. Davidson AH, Traub-Dargatz JL, Rodeheaver RM, et al.
likely induces protection against BoHV-1 in these animals. Immunologic responses to West Nile virus in vaccinated
10 LANGELLOTTI ET AL.

and clinically affected horses. J Am Vet Med Assoc 2005; cination against human immunodeficiency virus type-1.
226:240–245. Immunology 1999;98:436–442.
16. Denis M, Slaoui M, Keil G, et al. Identification of different 35. Jones C, and Chowdhury S. A review of the biology of
target glycoproteins for bovine herpes virus type 1-specific bovine herpesvirus type 1 (BHV-1), its role as a cofactor in
cytotoxic T lymphocytes depending on the method of the bovine respiratory disease complex and development of
in vitro stimulation. Immunology 1993;78:7–13. improved vaccines. Anim Health Res Rev 2007;8:187–205.
17. Deshpande MS, Ambagala TC, Hegde NR, et al. Induction 36. Jones C, and Chowdhury S. Bovine herpesvirus type 1
of cytotoxic T-lymphocytes specific for bovine herpesvirus- (BHV-1) is an important cofactor in the bovine respiratory
1 by DNA immunization. Vaccine 2002;20:3744–3751. disease complex. Vet Clin North Am Food Anim Pract
18. Di Giacomo S, Quattrocchi V, and Zamorano P. Use of 2010;26:303–321
adjuvants to enhance the immune response induced by a 37. Klaus GG, Pepys MB, Kitajima K, et al. Activation of
DNA vaccine against bovine herpesvirus-1. Viral Immunol mouse complement by different classes of mouse antibody.
2015;28:343–346. Immunology 1979;38:687–695.
19. Dupuis M, McDonald DM, and Ott G. Distribution of ad- 38. Kuhn JE, Kramer MD, Willenbacher W, et al. Identifica-
juvant MF59 and antigen gD2 after intramuscular injection tion of herpes simplex virus type 1 glycoproteins interact-
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

in mice. Vaccine 1999;18:434–439. ing with the cell surface. J Virol 1990;64:2491–2497.
20. Elgueta R, Benson MJ, de Vries VC, et al. Molecular 39. Langellotti C, Cesar G, Soria I, et al. Foot-and-mouth
mechanism and function of CD40/CD40L engagement in disease virus infection of dendritic cells triggers phos-
the immune system. Immunol Rev 2009;229:152–172. phorylation of ERK1/2 inducing class I presentation and
21. Ellis JA. Update on viral pathogenesis in BRD. Anim apoptosis. Vaccine 2015;33:4945–4953.
Health Res Rev 2009;10:149–153. 40. Langellotti CA, Pappalardo JS, Quattrocchi V, et al. In-
22. Flores-Romo L, Bjorck P, Duvert V, et al. CD40 ligation duction of specific cytotoxic activity for bovine
on human cord blood CD34+ hematopoietic progenitors herpesvirus-1 by DNA immunization with different adju-
induces their proliferation and differentiation into func- vants. Antiviral Res 2011;90:134–142.
tional dendritic cells. J Exp Med 1997;185:341–349. 41. Lewis PJ, Cox GJ, van Drunen Littel-van den Hurk S, et al.
23. Fooks AR, Jeevarajah D, Warnes A, et al. Immunization of Polynucleotide vaccines in animals: enhancing and modu-
mice with plasmid DNA expressing the measles virus nu- lating responses. Vaccine 1997;15:861–864.
cleoprotein gene. Viral Immunol 1996;9:65–71. 42. Lewis PJ, van Drunen Littel-van den H, and Babiuk LA.
24. Fuller AL, and Lee WC. Herpes simplex virus type 1 entry Altering the cellular location of an antigen expressed by a
through a cascade of virus-cell interactions requires different DNA-based vaccine modulates the immune response.
roles of gD and gH in penetration. J Virol 1992;66:5002–5012. J Virol 1999;73:10214–10223.
25. Galdiero S, Vitiello M, D’Isanto M, et al. The identification 43. Liang XP, Babiuk LA, van Drunen Littel-van den Hurk S,
and characterization of fusogenic domains in herpes virus et al. Bovine herpesvirus 1 attachment to permissive cells is
glycoprotein B molecules. Chembiochem 2008;9:758–767. mediated by its major glycoproteins gI, gIII, and gIV.
26. Garver KA, LaPatra SE, and Kurath G. Efficacy of an in- J Virol 1991;65:1124–1132.
fectious hematopoietic necrosis (IHN) virus DNA vaccine 44. Lodmell DL, Niwa A, Hayashi K, et al. Prevention of cell-
in Chinook Oncorhynchus tshawytscha and sockeye O. to-cell spread of herpes simplex virus by leukocytes. J Exp
nerka salmon. Dis Aquat Organ 2005;64:13–22. Med 1973;137:706–720.
27. Grewal IS, and Flavell RA. CD40 and CD154 in cell- 45. Lohr J, Knoechel B, Caretto D, et al. Balance of Th1 and
mediated immunity. Annu Rev Immunol 1998;16:111–135. Th17 effector and peripheral regulatory T cells. Microbes
28. Gurunathan S, Klinman DM, and Seder RA. DNA vac- Infect 2009;11:589–593.
cines: immunology, application, and optimization*. Annu 46. Manoj S, Babiuk LA, and van Drunen Littel-van den Hurk
Rev Immunol 2000;18:927–974. S. Immunization with a dicistronic plasmid expressing a
29. Hamilton JA, Byrne R, and Whitty G. Particulate adjuvants truncated form of bovine herpesvirus-1 glycoprotein D and
can induce macrophage survival, DNA synthesis, and a the amino-terminal subunit of glycoprotein B results in reduced
synergistic proliferative response to GM-CSF and CSF-1. gB-specific immune responses. Virology 2003;313:296–307.
J Leukoc Biol 2000;67:226–232. 47. Manoj S, Griebel PJ, Babiuk LA, et al. Targeting with bovine
30. Harcourt JL, Brown MP, Anderson LJ, et al. CD40 ligand CD154 enhances humoral immune responses induced by a
(CD154) improves the durability of respiratory syncytial DNA vaccine in sheep. J Immunol 2003;170:989–996.
virus DNA vaccination in BALB/c mice. Vaccine 2003;21: 48. Manoj S, Griebel PJ, Babiuk LA, et al. Modulation of
2964–2979. immune responses to bovine herpesvirus-1 in cattle by
31. Hariharan MJ, Nataraj C, and Srikumaran S. Down regu- immunization with a DNA vaccine encoding glycoprotein
lation of murine MHC class I expression by bovine her- D as a fusion protein with bovine CD154. Immunology
pesvirus 1. Viral Immunol 1993;6:273–284. 2004;112:328–338.
32. Huang Y, Babiuk LA, and van Drunen Littel-van den Hurk 49. Martins LP, Lau LL, Asano MS, et al. DNA vaccination against
S. Immunization with a bovine herpesvirus 1 glycoprotein persistent viral infection. J Virol 1995;69:2574–2582.
B DNA vaccine induces cytotoxic T-lymphocyte responses 50. Matzinger P. The JAM test. A simple assay for DNA
in mice and cattle. J Gen Virol 2005;86:887–898. fragmentation and cell death. J Immunol Methods 1991;
33. Hutchings DL, van Drunen Littel-van den Hurk S, and 145:185–192.
Babiuk LA. Lymphocyte proliferative responses to sepa- 51. Mendoza RB, Cantwell MJ, and Kipps TJ. Immunostimulatory
rated bovine herpesvirus 1 proteins in immune cattle. effects of a plasmid expressing CD40 ligand (CD154) on gene
J Virol 1990;64:5114–5122. immunization. J Immunol 1997;159:5777–5781.
34. Ihata A, Watabe S, Sasaki S, et al. Immunomodulatory 52. Miethke A, Keil GM, Weiland F, et al. Unidirectional
effect of a plasmid expressing CD40 ligand on DNA vac- complementation between glycoprotein B homologues of
CYTOTOXIC ACTIVITY USING A DNA VACCINE 11

pseudorabies virus and bovine herpesvirus 1 is determined 72. Stevens TL, Bossie A, Sanders VM, et al. Regulation of
by the carboxy-terminal part of the molecule. J Gen Virol antibody isotype secretion by subsets of antigen-specific
1995;76 (Pt 7):1623–1635. helper T cells. Nature 1988;334:255–258.
53. Mosmann TR, and Coffman RL. TH1 and TH2 cells: dif- 73. Tikoo SK, Campos M, and Babiuk LA. Bovine herpesvirus
ferent patterns of lymphokine secretion lead to different 1 (BHV-1): biology, pathogenesis, and control. Adv Virus
functional properties. Annu Rev Immunol 1989;7:145–173. Res 1995;45:191–223.
54. Muylkens B, Thiry J, Kirten P, Schynts F, and Thiry E. 74. Tikoo SK, Fitzpatrick DR, Babiuk LA, et al. Molecular
2007, Bovine herpesvirus 1 infection and infectious bovine cloning, sequencing, and expression of functional bovine
rhinotracheitis. Vet Res 1989;38:181–209. herpesvirus 1 glycoprotein gIV in transfected bovine cells.
55. Nataraj C, Eidmann S, Hariharan MJ, et al. Bovine her- J Virol 1990;64:5132–5142.
pesvirus 1 downregulates the expression of bovine MHC 75. van Drunen Littel-van den H, Braun RP, Lewis PJ, et al.
class I molecules. Viral Immunol 1997;10:21–34. Intradermal immunization with a bovine herpesvirus-1
56. Nguyen L, Knipe DM, and Finberg RW. Mechanism of virus- DNA vaccine induces protective immunity in cattle. J Gen
induced Ig subclass shifts. J Immunol 1994;152:478–484. Virol 1998;79 (Pt 4):831–839.
57. Okazaki K, Matsuzaki T, Sugahara Y, et al. BHV-1 ad- 76. van Drunen Littel-van den Hurk S, G G, and Babiuk LA.
Downloaded by AUT UNIVERSITY (Auckland University of Tech) from www.liebertpub.com at 11/05/20. For personal use only.

sorption is mediated by the interaction of glycoprotein gIII Epitope specificity of the protective immune response in-
with heparinlike moiety on the cell surface. Virology 1991; duced by individual bovine herpesvirus-1 glycoproteins.
181:666–670. Vaccine 1990;8:358–368
58. Oliveira SC, Harms JS, Rosinha GM, et al. Biolistic- 77. van Drunen Littel-van den Hurk S, Parker MD, Massie B, et al.
mediated gene transfer using the bovine herpesvirus-1 Protection of cattle from BHV-1 infection by immunization
glycoprotein D is an effective delivery system to induce with recombinant glycoprotein gIV. Vaccine 1993;11:25–35.
neutralizing antibodies in its natural host. J Immunol 78. Vialle R, Dupuis L, Deville S, et al. Microgel particulate
Methods 2000;245:109–118. adjuvant: characterization and mechanisms of action. Pro-
59. Ormsbee R. IBR and abortions. California Vet 1963;17:23–28. cedia Vaccinol 2010;2:12–16.
60. Pardoll DM, and Beckerleg AM. Exposing the immunology 79. Whetstone CA, Miller JM, Seal BS, et al. Latency and
of naked DNA vaccines. Immunity 1995;3:165–169. reactivation of a thymidine kinase-negative bovine her-
61. Quattrocchi V, Soria I, Langellotti CA, et al. A DNA pesvirus 1 deletion mutant. Arch Virol 1992;122:207–214.
vaccine formulated with chemical adjuvant provides partial 80. Whetstone CA, Wheeler JG, and Reed DE. Investigation of
protection against bovine herpes virus infection in cattle. possible vaccine-induced epizootics of infectious bovine
Front Immunol 2017;8:37. rhinotracheitis, using restriction endonuclease analysis of
62. Raaperi K, Orro T, and Viltrop A. Epidemiology and viral DNA. Am J Vet Res 1986;47:1789–1795.
control of bovine herpesvirus 1 infection in Europe. Vet J 81. Wienhold D, Armengol E, Marquardt A, et al. Im-
2014;201:249–256. munomodulatory effect of plasmids co-expressing cyto-
63. Raz E, Tighe H, Sato Y, et al. Preferential induction of a kines in classical swine fever virus subunit gp55/E2-DNA
Th1 immune response and inhibition of specific IgE anti- vaccination. Vet Res 2005;36:571–587.
body formation by plasmid DNA immunization. Proc Natl 82. Wyler R, Engels M, and Schwyzer M. Infectious bovine
Acad Sci U S A 1996;93:5141–5145. rhinotracheitis/vulvovaginitis (BHV-l). In: Herpesvirus Dis-
64. Ridge JP, Di Rosa F, and Matzinger P. A conditioned eases of Cattle, Horses, and Pigs, Wittmann G (ed.). Kluwer
dendritic cell can be a temporal bridge between a CD4+ Academic Publishers, Boston/Dordrecht/London, 1989.
T-helper and a T-killer cell. Nature 1998;393:474–478. 83. Xu S, and Cao X. Interleukin-17 and its expanding bio-
65. Romera SA, Puntel M, Quattrocchi V, et al. Protection logical functions. Cell Mol Immunol 2010;7:164–174.
induced by a glycoprotein E-deleted bovine herpesvirus
type 1 marker strain used either as an inactivated or live Address correspondence to:
attenuated vaccine in cattle. BMC Vet Res 2014;10:8. Dr. Patricia I. Zamorano
66. Roy M, Waldschmidt T, Aruffo A, et al. The regulation of Instituto de Virologı́a-IVIT
the expression of gp39, the CD40 ligand, on normal and (INTA-CONICET)
cloned CD4+ T cells. J Immunol 1993;151:2497–2510. De las Cabañas y De Los Reseros s/n
67. Schiffelers MJ, Blaauboer B, Bakker W, et al. Replacing Hurlingham, Buenos Aires, 1686
the NIH test for rabies vaccine potency testing: a synopsis Argentina
of drivers and barriers. Biologicals 2014;42:205–217.
Buenos Aires
68. Schoenberger SP, Toes RE, van der Voort EI, et al. T-cell
Argentina
help for cytotoxic T lymphocytes is mediated by CD40-
CD40L interactions. Nature 1998;393:480–483. E-mail: zamorano.patricia@inta.gob.ar
69. Sin JI, Kim JJ, Zhang D, et al. Modulation of cellular re-
sponses by plasmid CD40L: CD40L plasmid vectors enhance Dr. Cecilia A. Langellotti
antigen-specific helper T cell type 1 CD4+ T cell-mediated Instituto de Virologı́a-IVIT
protective immunity against herpes simplex virus type 2 (INTA-CONICET)
in vivo. Hum Gene Ther 2001;12:1091–1102. De las Cabañas y De Los Reseros s/n
70. Smith KC. Herpesviral abortion in domestic animals. Vet J Hurlingham, Buenos Aires, 1686
1997;153:253–268. Argentina
71. Socha W, Rola J, Urban-Chmiel R, et al. Application of Buenos Aires
loop-mediated isothermal amplification (LAMP) assays for Argentina
the detection of bovine herpesvirus 1. Pol J Vet Sci 2017;
20:619–622. E-mail: langellotti.cecilia@inta.gob.ar

You might also like